Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 10(7): 721-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19503103

ABSTRACT

Activation of the complement system generates potent chemoattractants and leads to the opsonization of cells for immune clearance. Short-lived protease complexes cleave complement component C3 into anaphylatoxin C3a and opsonin C3b. Here we report the crystal structure of the C3 convertase formed by C3b and the protease fragment Bb, which was stabilized by the bacterial immune-evasion protein SCIN. The data suggest that the proteolytic specificity and activity depend on the formation of dimers of C3 with C3b of the convertase. SCIN blocked the formation of a productive enzyme-substrate complex. Irreversible dissociation of the complex of C3b and Bb is crucial to complement regulation and was determined by slow binding kinetics of the Mg(2+)-adhesion site in Bb. Understanding the mechanistic basis of the central complement-activation step and microbial immune evasion strategies targeting this step will aid in the development of complement therapeutics.


Subject(s)
Bacterial Proteins/chemistry , Complement C3 Convertase, Alternative Pathway/chemistry , Complement Inactivator Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Binding Sites , Catalysis , Catalytic Domain , Complement C3/chemistry , Complement C3/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Complement C3-C5 Convertases/chemistry , Complement C3-C5 Convertases/metabolism , Complement C3b/chemistry , Complement C3b/metabolism , Complement Inactivator Proteins/immunology , Complement Inactivator Proteins/metabolism , Complement Pathway, Alternative/immunology , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Staphylococcus aureus/chemistry , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Substrate Specificity , Surface Plasmon Resonance
2.
Pediatr Nephrol ; 35(1): 153-162, 2020 01.
Article in English | MEDLINE | ID: mdl-31667615

ABSTRACT

BACKGROUND: C3 glomerulopathy (C3G) is defined by dominant glomerular deposition of C3 and minimal or no immunoglobulin, with two subtypes-dense deposit disease (DDD) and C3 glomerulonephritis (C3GN)-distinguished by features on electron microscopy (EM). Given that this rare disease has generally unfavorable yet highly variable outcomes, we sought out to review the histopathology, complement/genetic studies, and renal outcomes of pediatric patients with C3G at our institution. METHODS: All native kidney biopsies performed in a single pediatric hospital over a 10-year period were reviewed for features of C3G. Of 589 biopsy reports, we identified 9 patients fulfilling the diagnostic criteria for C3G and retrospectively reviewed their clinical chart and renal biopsy findings. RESULTS: We identified 4 patients with DDD, 4 with C3GN, and 1 indeterminate case, with features of both C3GN and DDD. Five patients were positive for one or more nephritic factors (C3NeF, C4NeF, C5NeF) with 1 patient additionally positive for complement factor H (CFH) autoantibody. Genetic testing done in 5 of the 9 patients failed to identify any causative mutations. Three patients showed progressive renal dysfunction over a mean follow-up period of 33 months. CONCLUSIONS: Complement and genetic studies are now routinely recommended for patients with a histopathological diagnosis of C3G. Careful interpretation of these studies and their prognostic and therapeutic implications in conjunction with biopsy findings is needed to further understand the pathophysiology of this rare disease in children.


Subject(s)
Complement C3/immunology , Glomerulonephritis, Membranoproliferative/immunology , Kidney Failure, Chronic/epidemiology , Kidney Glomerulus/pathology , Adolescent , Biopsy , Child , Complement C3/genetics , Complement Inactivator Proteins/analysis , Complement Inactivator Proteins/immunology , Disease Progression , Female , Follow-Up Studies , Genetic Testing , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/complications , Glomerulonephritis, Membranoproliferative/genetics , Humans , Kidney Failure, Chronic/immunology , Kidney Glomerulus/immunology , Kidney Glomerulus/ultrastructure , Male , Microscopy, Electron , Mutation , Retrospective Studies
3.
Nature ; 478(7367): 76-81, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21979047

ABSTRACT

Oxidative stress and enhanced lipid peroxidation are linked to many chronic inflammatory diseases, including age-related macular degeneration (AMD). AMD is the leading cause of blindness in Western societies, but its aetiology remains largely unknown. Malondialdehyde (MDA) is a common lipid peroxidation product that accumulates in many pathophysiological processes, including AMD. Here we identify complement factor H (CFH) as a major MDA-binding protein that can block both the uptake of MDA-modified proteins by macrophages and MDA-induced proinflammatory effects in vivo in mice. The CFH polymorphism H402, which is strongly associated with AMD, markedly reduces the ability of CFH to bind MDA, indicating a causal link to disease aetiology. Our findings provide important mechanistic insights into innate immune responses to oxidative stress, which may be exploited in the prevention of and therapy for AMD and other chronic inflammatory diseases.


Subject(s)
Complement Factor H/metabolism , Epitopes/metabolism , Malondialdehyde/metabolism , Oxidative Stress , Animals , Apoptosis , Binding Sites/genetics , Complement Factor H/genetics , Complement Factor H/immunology , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/immunology , Complement Inactivator Proteins/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipid Peroxidation , Macrophages, Peritoneal/metabolism , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Malondialdehyde/antagonists & inhibitors , Malondialdehyde/chemistry , Malondialdehyde/immunology , Mice , Mice, Inbred C57BL , Mutation/genetics , Necrosis , Protein Binding/genetics , Protein Structure, Tertiary , Retina/metabolism
4.
J Immunol ; 188(9): 4450-9, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22467648

ABSTRACT

Ail is a 17-kDa chromosomally encoded outer membrane protein that mediates serum resistance (complement resistance) in the pathogenic Yersiniae (Yersinia pestis, Y. enterocolitica, and Y. pseudotuberculosis). In this article, we demonstrate that Y. pseudotuberculosis Ail from strains PB1, 2812/79, and YPIII/pIB1 (serotypes O:1a, O:1b, and O:3, respectively) can bind the inhibitor of the classical and lectin pathways of complement, C4b-binding protein (C4BP). Binding was observed irrespective of serotype tested and independently of YadA, which is the primary C4BP receptor of Y. enterocolitica. Disruption of the ail gene in Y. pseudotuberculosis resulted in loss of C4BP binding. Cofactor assays revealed that bound C4BP is functional, because bound C4BP in the presence of factor I cleaved C4b. In the absence of YadA, Ail conferred serum resistance to strains PB1 and YPIII, whereas serum resistance was observed in strain 2812/79 in the absence of both YadA and Ail, suggesting additional serum resistance factors. Ail from strain YPIII/pIB1 alone can mediate serum resistance and C4BP binding, because its expression in a serum-sensitive laboratory strain of Escherichia coli conferred both of these phenotypes. Using a panel of C4BP mutants, each deficient in a single complement control protein domain, we observed that complement control protein domains 6-8 are important for binding to Ail. Binding of C4BP was unaffected by increasing heparin or salt concentrations, suggesting primarily nonionic interactions. These results indicate that Y. pseudotuberculosis Ail recruits C4BP in a functional manner, facilitating resistance to attack from complement.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Complement Inactivator Proteins/immunology , Complement Pathway, Classical/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , Histocompatibility Antigens/immunology , Yersinia pseudotuberculosis/immunology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Adhesins, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Blood Bactericidal Activity/genetics , Blood Bactericidal Activity/immunology , Complement C1/genetics , Complement C1/immunology , Complement C1/metabolism , Complement C4b-Binding Protein , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/metabolism , Complement Pathway, Classical/genetics , Complement Pathway, Mannose-Binding Lectin/genetics , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli/metabolism , Female , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Humans , Male , Mutation , Protein Binding/genetics , Protein Binding/immunology , Species Specificity , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/metabolism
5.
Trends Immunol ; 30(6): 286-92, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19428302

ABSTRACT

Several studies of human cancers have established that chronic and insidious inflammation promotes the process of carcinogenesis and exacerbates the growth of existing tumors. Conversely, acute inflammation seems to have the opposite effect. Recent discoveries indicate that this dualism in the role of inflammation in cancer is mirrored by the effects of the complement system on this disease process. Previous studies have suggested that complement proteins can contribute to the immune surveillance of malignant tumors. However, a very recent study has indicated that complement proteins can also promote tumor growth. Here, we describe our current understanding of the role of complement in tumor development and progression.


Subject(s)
Complement Inactivator Proteins/immunology , Complement System Proteins/immunology , Neoplasms/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Complement Inactivator Proteins/metabolism , Complement System Proteins/metabolism , Disease Progression , Humans , Immunologic Surveillance , Inflammation , Neoplasms/pathology , Oxidative Stress , Risk Factors , Tumor Escape
6.
Adv Exp Med Biol ; 946: 113-33, 2012.
Article in English | MEDLINE | ID: mdl-21948365

ABSTRACT

Our understanding of both the nature and diversity of Staphylococcal immune evasion proteins has increased tremendously throughout the last several years. Among this group of molecules, members of the SCIN and Efb families of complement inhibitors have been the subject of particularly intense study. This work has demonstrated that both types of proteins exert their primary function by inhibiting C3 convertases, which lie at the heart of the complement-mediated immune response. Despite this similarity, however, significant differences in structure/function relationships and mechanisms of action exist between these bacterial proteins. Furthermore, divergent secondary effects on host immune responses have also been described for these two protein families. This chapter summarizes recent advances toward understanding the structure, function, and mechanism of the SCIN and Efb families, and suggests potential directions for the field over the coming years.


Subject(s)
Bacterial Proteins/immunology , Complement C3b Inactivator Proteins/immunology , Complement Inactivator Proteins/immunology , Immunity, Innate/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Bacterial Proteins/chemistry , Complement C3b Inactivator Proteins/chemistry , Complement Inactivator Proteins/chemistry , Humans , Staphylococcal Infections/microbiology , Structure-Activity Relationship
7.
Am J Pathol ; 177(6): 3224-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20971742

ABSTRACT

Inflammation and activation of the complement system predispose to intracranial artery aneurysm (IA) rupture. Because disturbances in complement regulation may lead to increased susceptibility to complement activation and inflammation, we looked for evidence for dysregulation of the complement system in 26 unruptured and 26 ruptured IAs resected intraoperatively. Immunohistochemical and immunofluorescence results of parallel IA sections showed that deposition of the complement activation end-product C5b-9 was lacking from the luminal part of the IA wall that contained complement inhibitors factor H, C4b binding protein, and protectin as well as glycosaminoglycans. In contrast, the outer, less cellular part of the IA wall lacked protectin and had enabled full complement activation and C5b-9 formation. Decay accelerating factor and membrane cofactor protein had less evident roles in complement regulation. The Factor H Y402H variant, studied in 97 IA patients, was seen as often in aneurysm patients with or without aneurysm rupture as in the control population. The regulatory capacity of the complement system thus appears disturbed in the outer part of the IA wall, allowing full proinflammatory complement activation to occur before aneurysm rupture. Insufficient complement control might be due to matrix remodeling and cell loss by mechanical hemodynamics and/or inflammatory stress. Apparently, disturbed complement regulation leads to an increased susceptibility to complement activation, inflammation, and tissue damage in the IA wall.


Subject(s)
Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Complement Inactivator Proteins/metabolism , Complement Pathway, Classical/immunology , Intracranial Aneurysm/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aneurysm, Ruptured/immunology , Aneurysm, Ruptured/metabolism , Aneurysm, Ruptured/pathology , Cerebral Arteries/immunology , Complement Activation/immunology , Complement Inactivator Proteins/immunology , Complement Pathway, Classical/physiology , Down-Regulation/immunology , Female , Humans , Intracranial Aneurysm/immunology , Intracranial Aneurysm/pathology , Male , Middle Aged , Young Adult
8.
Biologicals ; 39(1): 38-42, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21256764

ABSTRACT

Aggregates in human immunoglobulin (Ig) products can develop due to employed manufacturing, formulation and storage conditions and can cause adverse reactions in patients. The test for anti-complementary activity (ACA) recommended by the European Pharmacopoeia (EP) is insensitive, variable and time consuming. We have optimised a commercial assay for the detection and quantitation of C1q binding aggregates in intravenous and intramuscular IgG preparations. The generation of C4d, iC3b and SC5b-9 induced by aggregates in vitro was measured by enzyme-linked immunosorbent assays (ELISA). In establishing the sensitivity of the C1q aggregate binding assay to detect IgG aggregates in comparison to turbidity and ACA, pure IgG at neutral and acidic pH was heated for various lengths of time to generate varying amounts of aggregates. The level of C1q binding aggregates was 7 fold greater in intramuscular samples. These aggregates were capable of activating complement in vitro and correlated with an increase in ACA. C1q aggregate binding was apparent before any quantifiable turbidity and ACA in the heat-treated samples. Furthermore, the C1q binding assay could discriminate between different levels of aggregates where ACA had reached a plateau. C1q aggregate binding is a sensitive, convenient, specific and robust means of detecting aggregates with a propensity for complement activation.


Subject(s)
Complement C1q/immunology , Complement Inactivator Proteins/immunology , Immunoglobulins/immunology , Complement Activation/immunology , Complement C1q/metabolism , Complement Inactivator Proteins/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/standards , Hot Temperature , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulins/metabolism , Immunoglobulins, Intravenous/immunology , Immunoglobulins, Intravenous/metabolism , Protein Binding/immunology , Reproducibility of Results
9.
J Invertebr Pathol ; 107 Suppl: S71-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21784233

ABSTRACT

Serum inactivation of baculovirus vectors is a significant barrier to the development of these highly efficient vectors for therapeutic gene delivery. In this review we will describe the efforts taken to avoid complement attack by passive or active measures. Evidently good targets for baculovirus-mediated gene delivery include immunoprivileged tissues, such as eye, brain and testis. Similarly baculovirus vectors have also proven their efficacy in an ex vivo setting for tissue engineering. Active measures to inhibit complement include the use of pharmacological inhibitors of complement as well as surface engineering of the baculoviral vectors through the use of synthetic polymers, pseudotyping or display of complement inhibitors. Lessons learned from these studies will significantly increase the possibility of using baculovirus vectors for therapeutic applications.


Subject(s)
Baculoviridae/immunology , Complement Activation , Complement Inactivator Proteins/immunology , Complement Membrane Attack Complex/immunology , Genetic Therapy/methods , Nucleopolyhedroviruses/immunology , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Complement Inactivator Proteins/therapeutic use , Complement Membrane Attack Complex/metabolism , Gene Transfer Techniques , Humans , Immunologic Factors , Nucleopolyhedroviruses/genetics
10.
Front Immunol ; 12: 732146, 2021.
Article in English | MEDLINE | ID: mdl-34707606

ABSTRACT

To assess the relative contribution of opsonisation by antibodies, classical and alternative complement pathways to pneumococcal phagocytosis, we analyzed killing of pneumococci by human blood leukocytes collected from vaccine-naïve and PCV13-vaccinated subjects. With serotype 4 pneumococci as model, two different physiologic opsonophagocytosis assays based on either hirudin-anticoagulated whole blood or on washed cells from EDTA-anticoagulated blood reconstituted with active serum, were compared. Pneumococcal killing was measured in the presence of inhibitors targeting the complement components C3, C5, MASP-2, factor B or factor D. The two assay formats yielded highly consistent and comparable results. They highlighted the importance of alternative complement pathway activation for efficient opsonophagocytic killing in blood of vaccine-naïve subjects. In contrast, alternative complement pathway inhibition did not affect pneumococcal killing in PCV13-vaccinated individuals. Independent of amplification by the alternative pathway, even low capsule-specific antibody concentrations were sufficient to efficiently trigger classical pathway mediated opsonophagocytosis. In heat-inactivated or C3-inhibited serum, high concentrations of capsule-specific antibodies were required to trigger complement-independent opsonophagocytosis. Our findings suggest that treatment with alternative complement pathway inhibitors will increase susceptibility for invasive pneumococcal infection in non-immune subjects, but it will not impede pneumococcal clearance in vaccinated individuals.


Subject(s)
Complement Pathway, Alternative , Complement System Proteins/immunology , Opsonization , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Streptococcus pneumoniae/immunology , Vaccination , Adult , Aged , Complement Inactivator Proteins/immunology , Complement Inactivator Proteins/metabolism , Complement System Proteins/metabolism , Female , Host-Pathogen Interactions , Humans , Longitudinal Studies , Male , Middle Aged , Pneumococcal Infections/blood , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/pathogenicity
11.
J Exp Med ; 154(1): 1-12, 1981 Jul 01.
Article in English | MEDLINE | ID: mdl-7019379

ABSTRACT

Three mechanisms that regulate the formation and function of the classical pathway C3 convertase (C4b2a) have been elucidated: (a) an intrinsic decay of the enzyme that is temperature dependent; (b) an extrinsic decay mediated by the effect of the serum protein C4b binding protein (C4-bp); and (c) inactivation of C4b by the proteolytic action of C4b/C3b inactivator (C4b/C3bINA), which cleaves that alpha' chain of C4b to yield C4d (alpha 2) and C4c (alpha 3, alpha 4, beta, and gamma chains). A fourth mechanism described here is based on the observation that the IgG fraction of the serum of certain patients with glomerulonephritis contains a protein termed C4 nephritic factor (NFc), which prevents the intrinsic decay of C4b2a. This protein, which prolongs the half-life of surface-bound C4b2a from 7.5 min to greater than 5 h, increases the use of C3 and C5. It also inhibits the decay produced by C4-bp by preventing the dissociation of C2a from the C4b2a complex. Additionally, the C2b/C3bINA alone, or in the presence of C4-bp, fails to cleave the alpha' chain of C4b in the surface-bound stabilized C4b2a complex. This protective property of NFc requires the presence of C2a, because C4b was not protected unless it was bound to C2a. Thus in the presence of NFc, the three natural controls of the function of the classical pathway convertase, intrinsic decay, extrinsic decay, and proteolytic cleavage, are bypassed.


Subject(s)
Complement Activating Enzymes/metabolism , Complement Activation , Complement C3 Nephritic Factor/immunology , Complement C3-C5 Convertases/metabolism , Complement C4/immunology , Complement Inactivator Proteins/immunology , Complement Pathway, Classical , Animals , Complement C2/metabolism , Complement C3/metabolism , Guinea Pigs , Humans , Peptide Hydrolases/pharmacology , Rabbits , Rats
12.
J Exp Med ; 186(7): 1015-26, 1997 Oct 06.
Article in English | MEDLINE | ID: mdl-9314551

ABSTRACT

Complement (C) is an important component of innate immunity, and was also shown recently to participate in induction of acquired B cell humoral immunity. In this study, we present evidence that C also participates in acquired T cell immunity. We found that C was involved in early events of the efferent elicitation phase of contact sensitivity (CS), and delayed-type hypersensitivity (DTH). Thus, CS and DTH were inhibited by administration of a C-blocker, soluble recombinant C receptor-1 (sCR1), when given 30 min before, but not 3 h after local antigen challenge. Among C components, local C5 were thought crucial to elicitation of CS, since local administration of anti-C5 monoclonal antibodies or locally injected C-depleting cobra venom factor also inhibited CS and DTH. These findings were consistent with our previous finding of the importance of C5 for CS elicitation, using congenitally C5-deficient mice. To dissect the mechanism of C dependence in CS, we demonstrated that locally increased early macrophage chemotactic activity (probably C5a) in evolving CS skin extracts, as well as late elaboration of IFN-gamma, were both inhibited by anti-C treatment. In addition, histological analysis showed that leukocyte recruitment into CS ear sites was similarly C-dependent. Furthermore, an initiating role of B cell-derived C-fixing immunoglobulin was suggested by demonstration of impaired CS responses in B cell-deficient mice. In summary, these results suggest that C was activated locally, perhaps via a B cell product, in an important early component of the stepwise events necessary to elicit CS, leading to local production of C5-dependent macrophage chemotactic activity and later IFN-gamma, and subsequently leading to cell infiltration, for development of T cell-dependent CS.


Subject(s)
B-Lymphocytes/immunology , Complement Activation/immunology , Complement C5/immunology , Dermatitis, Contact/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Chemotactic Factors/biosynthesis , Chemotaxis , Complement C5/metabolism , Complement Inactivator Proteins/immunology , Complement Inactivator Proteins/pharmacology , Elapid Venoms/pharmacology , Female , Hypersensitivity, Delayed/immunology , Interferon-gamma/biosynthesis , Macrophages/physiology , Mice , Mice, Inbred Strains , Receptors, Complement/immunology , Recombinant Proteins/pharmacology , Skin/immunology , T-Lymphocytes/metabolism
13.
J Exp Med ; 162(1): 75-92, 1985 Jul 01.
Article in English | MEDLINE | ID: mdl-2409211

ABSTRACT

Decay-accelerating factor (DAF) is a 70,000 Mr protein that has been isolated from the membrane of red cells. The function of DAF is to inhibit the assembly of amplifying enzymes of the complement cascade on the cell surface, thereby protecting them from damage by autologous complement. We raised monoclonal antibodies to DAF and used them to study its distribution in cells from the peripheral blood of normal individuals and of patients with paroxysmal nocturnal hemoglobinuria (PNH), a disease characterized by the unusual susceptibility of red cells to the hemolytic activity of complement. The results of immunoradiometric assays and of fluorescence-activated cell sorter analysis showed that DAF was present not only on red cells but was widely distributed on the surface membrane of platelets, neutrophils, monocytes, and B and T lymphocytes. By Western blotting, we observed small but consistent differences in the Mr of DAF from the membranes of various cell types. Quantitative studies showed that phagocytes and B lymphocytes, which presumably enter more frequently in contact with immune complexes and other potential activators of complement, had the highest DAF levels. As previously reported by others, the red cells from PNH patients were DAF deficient. When the patients' red cells were incubated in acidified serum (Ham test), only the DAF-deficient cells were lysed. In addition, we detected defects in DAF expression on platelets and all types of leukocytes. The observed patterns of DAF deficiency in these patients were consistent with the concept that the PNH cells were of monoclonal origin. In one patient, abnormal and normal cells were found only in the erythroid, myeloid, and megakaryocytic lineages. In two other patients, the lymphocytes were also DAF deficient, suggesting that a mutation occurred in a totipotent stem cell. It appears, therefore, that the lesion leading to PNH can occur at various stages in the differentiation of hematopoietic cells.


Subject(s)
Blood Proteins/analysis , Complement Inactivator Proteins/blood , Hemoglobinuria, Paroxysmal/blood , Antibodies, Monoclonal/immunology , Blood Platelets/metabolism , Blood Proteins/deficiency , Blood Proteins/immunology , CD55 Antigens , Complement Inactivator Proteins/deficiency , Complement Inactivator Proteins/immunology , Erythrocyte Membrane/immunology , Erythrocyte Membrane/metabolism , Hemoglobinuria, Paroxysmal/immunology , Humans , Leukocytes/metabolism
14.
J Exp Med ; 194(11): 1609-16, 2001 Dec 03.
Article in English | MEDLINE | ID: mdl-11733575

ABSTRACT

We studied complement 1 inhibitor (C1-INH) as an inhibitor of the alternative complement pathway. C1-INH prevented lysis, induced by the alternative complement pathway, of paroxysmal nocturnal hemoglobinuria (PNH) erythrocytes in human serum. It inhibited the binding of both factors B and C3 to PNH and rabbit erythrocytes and blocked the ability of factor B to restore alternative-pathway function in factor B-depleted serum. C1-INH did not bind to factors B or D but did bind to immobilized C3b and cobra venom factor (CVF), a C3b analogue. C1-INH prevented factor B from binding to CVF-coated beads and dissociated bound factor B from such beads. Factor B and C1-INH showed cross competition in binding to CVF-coated beads. Factor D cleaved factor B into Bb and Ba in the presence of C3b. Cleavage was markedly inhibited when C3b was preincubated with C1-INH. C1-INH inhibited the formation of CVFBb and decreased the C3 cleavage. Removal of C1-INH from serum, in the presence of Mg-EGTA with an anti-C1-INH immunoabsorbant, markedly increased alternative-pathway lysis. C1-INH interacts with C3b to inhibit binding of factor B to C3b. At physiologic concentrations, it is a downregulator of the alternative pathway convertase.


Subject(s)
Complement C1 Inactivator Proteins/immunology , Complement C3b/immunology , Complement Factor B/immunology , Complement Factor D/immunology , Complement Pathway, Alternative/immunology , Glycoproteins/immunology , Hemoglobinuria, Paroxysmal/immunology , Absorption , Animals , Complement C1 Inactivator Proteins/metabolism , Complement C1 Inactivator Proteins/pharmacology , Complement C3 Convertase, Alternative Pathway , Complement C3b/metabolism , Complement Factor B/metabolism , Complement Factor D/metabolism , Complement Inactivator Proteins/immunology , Complement Inactivator Proteins/metabolism , Egtazic Acid/pharmacology , Elapid Venoms/immunology , Elapid Venoms/metabolism , Erythrocytes/drug effects , Erythrocytes/immunology , Erythrocytes/metabolism , Glycoproteins/metabolism , Glycoproteins/pharmacology , Hemoglobinuria, Paroxysmal/blood , Hemolysis/drug effects , Hemolysis/immunology , Humans , Microspheres , Peptide Fragments/metabolism , Rabbits , Sepharose
15.
Ann Neurol ; 65(1): 67-75, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19194881

ABSTRACT

OBJECTIVE: Complement mediated injury of the neuromuscular junction is considered a primary disease mechanism in human myasthenia gravis and animal models of experimentally acquired myasthenia gravis (EAMG). We utilized active and passive models of EAMG to investigate the efficacy of a novel C5 complement inhibitor rEV576, recombinantly produced protein derived from tick saliva, in moderating disease severity. METHODS: Standardized disease severity assessment, serum complement hemolytic activity, serum cytotoxicity, acetylcholine receptor (AChR) antibody concentration, IgG subclassification, and C9 deposition at the neuromuscular junction were used to assess the effect of complement inhibition on EAMG induced by administration of AChR antibody or immunization with purified AChR. RESULTS: Administration of rEV576 in passive transfer EAMG limited disease severity as evidenced by 100% survival rate and a low disease severity score. In active EAMG, rats with severe and mild EAMG were protected from worsening of disease and had limited weight loss. Serum complement activity (CH(50)) in severe and mild EAMG was reduced to undetectable levels during treatment, and C9 deposition at the neuromuscular junction was reduced. Treatment with rEV576 resulted in reduction of toxicity of serum from severe and mild EAMG rats. Levels of total AChR IgG, and IgG(2a) antibodies were similar, but unexpectedly, the concentration of complement fixing IgG(1) antibodies was lower in a group of rEV576-treated animals, suggesting an effect of rEV576 on cellular immunity. INTERPRETATION: Inhibition of complement significantly reduced weakness in two models of EAMG. C5 inhibition could prove to be of significant therapeutic value in human myasthenia gravis.


Subject(s)
Complement C5/antagonists & inhibitors , Complement Inactivator Proteins/therapeutic use , Myasthenia Gravis, Autoimmune, Experimental/prevention & control , Animals , Antibodies/adverse effects , Arthropod Proteins , Cell Line, Tumor , Complement C9/metabolism , Complement Inactivator Proteins/immunology , Complement Inactivator Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Immunoglobulin G/blood , Immunoglobulin G/classification , Insect Proteins/immunology , Insect Proteins/therapeutic use , Muscle Strength/drug effects , Myasthenia Gravis, Autoimmune, Experimental/etiology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/pathology , Neuromuscular Junction/drug effects , Neuromuscular Junction/metabolism , Rats , Rats, Inbred Lew , Receptors, Cholinergic/immunology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Salivary Proteins and Peptides/blood , Salivary Proteins and Peptides/immunology , Salivary Proteins and Peptides/therapeutic use , Severity of Illness Index , Time Factors , Weight Loss/drug effects
16.
J Immunol ; 181(11): 7463-7, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017934

ABSTRACT

Staphylococcus aureus expresses a highly diversified arsenal of immune evasion proteins, many of which target the complement system. The extracellular fibrinogen-binding protein (Efb) and the Efb homologous protein (Ehp) have previously been demonstrated to bind to C3 and inhibit complement activation and amplification. In this study we present the first evidence that Efb and Ehp are also capable of inhibiting the interaction of C3d with complement receptor 2 (CR2), which plays an important role in B cell activation and maturation. The C-terminal domain of Efb efficiently blocked this interaction both in surface plasmon resonance-based competition studies and cellular assays and prevented the CR2-mediated stimulation of B cells. Furthermore, analyses of the available structural data were consistent with a molecular mechanism that reflects both steric and electrostatic effects on the C3d-CR2 interaction. Our study therefore suggests that S. aureus may disrupt both the innate and adaptive immune responses with a single protein module.


Subject(s)
Bacterial Proteins/immunology , Complement C3d/immunology , Complement Inactivator Proteins/immunology , Immunity, Innate , Receptors, Complement 3d/immunology , Staphylococcus aureus/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Proteins/metabolism , Cell Line , Complement Activation/immunology , Complement C3d/metabolism , Complement Inactivator Proteins/metabolism , Humans , Lymphocyte Activation/immunology , Mice , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Complement 3d/metabolism , Staphylococcus aureus/metabolism , Static Electricity , Structure-Activity Relationship , Surface Plasmon Resonance
17.
Nat Rev Neurol ; 16(11): 601-617, 2020 11.
Article in English | MEDLINE | ID: mdl-33005040

ABSTRACT

The complement system consists of a network of plasma and membrane proteins that modulate tissue homeostasis and contribute to immune surveillance by interacting with the innate and adaptive immune systems. Dysregulation, impairment or inadvertent activation of complement components contribute to the pathogenesis of some autoimmune neurological disorders and could even contribute to neurodegenerative diseases. In this Review, we summarize current knowledge about the main functions of the complement pathways and the involvement of complement in neurological disorders. We describe the complex network of complement proteins that target muscle, the neuromuscular junction, peripheral nerves, the spinal cord or the brain and discuss the autoimmune mechanisms of complement-mediated myopathies, myasthenia, peripheral neuropathies, neuromyelitis and other CNS disorders. We also consider the emerging role of complement in some neurodegenerative diseases, such as Alzheimer disease, amyotrophic lateral sclerosis and even schizophrenia. Finally, we provide an overview of the latest complement-targeted immunotherapies including monoclonal antibodies, fusion proteins and peptidomimetics that have been approved, that are undergoing phase I-III clinical trials or that show promise for the treatment of neurological conditions that respond poorly to existing immunotherapies.


Subject(s)
Complement Inactivating Agents/administration & dosage , Complement System Proteins/immunology , Drug Delivery Systems/trends , Immunotherapy/trends , Nervous System Diseases/immunology , Nervous System Diseases/therapy , Animals , Complement Inactivator Proteins/administration & dosage , Complement Inactivator Proteins/immunology , Complement System Proteins/metabolism , Drug Delivery Systems/methods , Humans , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/immunology , Immunotherapy/methods , Signal Transduction/drug effects , Signal Transduction/immunology , Synapses/drug effects , Synapses/immunology
18.
Front Immunol ; 10: 752, 2019.
Article in English | MEDLINE | ID: mdl-31024572

ABSTRACT

The interactions of cancer cells with components of the complement system are highly complex, leading to an outcome that is either favorable or detrimental to cancer cells. Currently, we perceive only the "tip of the iceberg" of these interactions. In this review, we focus on the complement terminal C5b-9 complex, known also as the complement membrane attack complex (MAC) and discuss the complexity of its interaction with cancer cells, starting with a discussion of its proposed mode of action in mediating cell death, and continuing with a portrayal of the strategies of evasion exhibited by cancer cells, and closing with a proposal of treatment approaches targeted at evasion strategies. Upon intense complement activation and membrane insertion of sufficient C5b-9 complexes, the afflicted cells undergo regulated necrotic cell death with characteristic damage to intracellular organelles, including mitochondria, and perforation of the plasma membrane. Several pro-lytic factors have been proposed, including elevated intracellular calcium ion concentrations and activated JNK, Bid, RIPK1, RIPK3, and MLKL; however, further research is required to fully characterize the effective cell death signals activated by the C5b-9 complexes. Cancer cells over-express a multitude of protective measures which either block complement activation, thus reducing the number of membrane-inserted C5b-9 complexes, or facilitate the elimination of C5b-9 from the cell surface. Concomitantly, cancer cells activate several protective pathways that counteract the death signals. Blockage of complement activation is mediated by the complement membrane regulatory proteins CD46, CD55, and CD59 and by soluble complement regulators, by proteases that cleave complement proteins and by protein kinases, like CK2, which phosphorylate complement proteins. C5b-9 elimination and inhibition of cell death signals are mediated by caveolin and dynamin, by Hsp70 and Hsp90, by the mitochondrial stress protein mortalin, and by the protein kinases PKC and ERK. It is conceivable that various cancers and cancers at different stages of development will utilize distinct patterns of these and other MAC resistance strategies. In order to enhance the impact of antibody-based therapy on cancer, novel precise reagents that block the most effective protective strategies will have to be designed and applied as adjuvants to the therapeutic antibodies.


Subject(s)
Complement Membrane Attack Complex/metabolism , Neoplasms/immunology , Animals , Calcium Signaling , Cell Death/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Complement Activation , Complement Inactivator Proteins/immunology , Complement Inactivator Proteins/metabolism , Cytotoxicity, Immunologic , Humans , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Models, Immunological , Neoplasms/metabolism , Neoplasms/pathology , Tumor Microenvironment/immunology
19.
Cancer Res ; 66(12): 6258-63, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778201

ABSTRACT

Neoplasms have developed strategies to protect themselves against the complement-mediated host immunity. Invasion- and metastasis-promoting membrane type-1 (MT1) matrix metalloproteinase (MMP) is strongly associated with many metastatic cancer types. The relative importance of the individual functions of MT1-MMP in metastasis was, however, unknown. We have now determined that the expression of murine MT1-MMP in murine melanoma B16F1 cells strongly increased the number of metastatic loci in the lungs of syngeneic C57BL/6 mice. In contrast, MT1-MMP did not affect the number of metastatic loci in complement-deficient C57BL/6-C3-/- mice. Our results indicated, for the first time, that the anticomplement activity of MT1-MMP played a significant role in promoting metastasis in vivo and determined the relative importance of the anticomplement activity in the total metastatic effect of this multifunctional proteolytic enzyme. We believe that our results shed additional light on the functions of MT1-MMP in cancer and clearly make this protease a promising drug target in metastatic malignancies.


Subject(s)
Complement C3/immunology , Complement Inactivator Proteins/immunology , Matrix Metalloproteinases/immunology , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/immunology , Animals , Complement C3/deficiency , Complement C3/genetics , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/metabolism , Fibrosarcoma/enzymology , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 14 , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases, Membrane-Associated , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Transfection
20.
J Clin Invest ; 96(5): 2348-56, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7593622

ABSTRACT

The kidney widely expresses membrane-associated complement regulatory proteins (membrane inhibitors of complement). The aim of this work was to evaluate the roles of these molecules in rat kidneys in vivo. To suppress functions of rat membrane inhibitors of complement, two mAbs, 512 and 6D1, were used. 5I2 and 6D1 inhibit functions of membrane inhibitors of complement at C3 level (rat Crry/p65) and C8/9 level (rat CD59), respectively. F(ab')2 fragment of 5I2 or 6D1 was perfused in the left kidneys, and perfusate was discarded from the renal vein. After perfusion, the left kidneys were connected to systemic circulation. In rats perfused with 5I2, mouse IgG was found in glomeruli, peritubular capillaries, vascular bundles, and tubules 15 min after recirculation. Binding of C3 and C5b-9 was evident in these areas. 1 d after perfusion with 5I2, cast formation, dilatation of tubular lumen, and tubular cell degeneration were observed. At day 4 through day 7, significant mononuclear cell infiltration and proximal tubule damage were observed. These changes were completely prevented by complement depletion. Rats perfused with 6D1 showed the binding of mouse IgG in the similar areas as 5I2, but C3 or C5b-9 deposition was not observed. Rats perfused with 6D1 or vehicle only did not show any pathology in the left kidneys. These results suggest that rat Crry/p65 plays protective roles against spontaneously occurring indiscriminate attack to tubulointerstitial tissues by autologous complement and that rat Crry/p65 is one of the important factors to maintain normal integrity of the kidney in rats.


Subject(s)
Antibodies, Monoclonal/immunology , Complement C3/immunology , Complement Inactivator Proteins/immunology , Kidney/immunology , Animals , Complement C3/analysis , Female , Immunoglobulin G/immunology , Kidney/pathology , Kidney Tubules/immunology , Kidney Tubules/pathology , Mice , Perfusion , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL